Sie sind auf Seite 1von 18

Review

Stress and anxiety across the lifespan: structural


plasticity and epigenetic regulation

The brain is the central organ of the bodys response to and perception of stress. Both the juvenile and
the adult brain show a significant capacity for lasting physiological, structural and behavioral plasticity as
a consequence of stress exposure. The hypothesis that epigenetic mechanisms might lie behind the lasting
effects of stress upon the brain has proven a fruitful one. In this review, we examine the growing literature
showing that stress has a direct impact on epigenetic marks at all life history stages thus far examined
and how, in turn, epigenetic mechanisms play a role in altering stress responsiveness, anxiety and brain

of
plasticity across the lifespan and beyond to succeeding generations. In addition, we will examine our own
recent findings that stress interacts with the epigenome to regulate the expression of transposable
elements in a regionally specific fashion, a finding with significant implications for a portion of the genome
which is tenfold larger than that occupied by the genes themselves.

ro
Keywords: adolescence n adulthood n aging n DNA methylation n early life n histone Richard G Hunter*1,2 &
modification n ncRNA Bruce S McEwen1
1
Harold & Margaret Milliken Hatch
Stressful experiences occur across the lifespan stimulation [7] , intense learning [8] , learning Laboratory of Neuroendocrinology,
rP
The Rockefeller University,
and have lasting effects on the brain, body and and practicing of motor skills [9] , and acute and 1230 York Ave., New York, NY 10065,
behavior [1] . Stress is a contributor to a variety chronic stressors [4] . Circulating hormones play USA
2
Department of Psychology, University
of disorders, including anxiety and mood dis- a role along with endogenous neurotransmitters of Massachusetts, Boston,
orders, but also diseases like heart disease and and trophic factors [4] . 100Morrissey Blvd, Boston, MA 02125,
USA
metabolic syndrome [2] . Stress also has the capac- In the hippocampus and medial prefrontal *Author for correspondence:
ho

ity to induce brain plasticity in a variety of ways, cortex, chronic stress causes dendrites of neu- richard.hunter@umb.edu
including neurogenesis, and the structure of rons to shrink and become shorter and less
neurons in stress-sensitive brain regions like the branched, with an accompanying reduction in
hippocampus, amygdala and prefrontal cortex synaptic input [10,11] . This reduces capabilities for
[3] . Given the potential of stress to alter physiol- nuanced cognitive function, memory and self-
ut

ogy and behavior in a lasting way, particularly in regulation. Glucocorticoids of the adrenal cortex
a terminally differentiated tissue like the brain, and excitatory amino acid transmitters in those
many researchers made the logical conclusion brain regions participate in the depolymerization
that the mechanisms behind the persistence of of the cytoskeleton and shrinkage of dendrites,
A

such stress-induced alterations might be epigen- by a process that is largely reversible at the end
etic in nature. This review will describe the work of stress, at least in the young adult brain [4,10] .
that has begun to support this hypothesis, and With aging, there is some loss of resilience that
examine how stress and epigenetics interact in is shrinkage of dendrites occurs, but recovery is
the brain across the lifespan and across genera- impaired [12] . In contrast to hippocampus and
tions (see Table1 and Figure 1 for a schema of the the medial prefrontal cortex, the basolateral
interactions of brain and behavioral plasticity amygdala and orbitofrontal cortex respond to
with epigenetic actors and stress). the same chronic stress by expanding dendrites
and increasing synaptic input, which leads to
Stress & brain plasticity increased anxiety, vigilance and aggressiveness
The brain possesses a life-long capacity for [13,14] . Early-life events also affect how these
reversible, structural plasticity, which includes brain regions develop, as will be discussed below.
turnover of synaptic connections, expansion Stress-induced changes in brain regions in
and contraction of dendritic trees and a lim- the animal models discussed above are mir-
ited amount of neurogenesis, particularly in rored by findings for the human brain that
the dentate gyrus of the hippocampus [4] . This include shrinkage of the hippocampus in pro-
plasticity can be seen as a result of an enriched longed major depression and Cushings disease,
environment [5] , regular exercise [6] , sensory as well as in post-traumatic stress disorder part of

10.2217/EPI.13.8 2013 Future Medicine Ltd Epigenomics (2013) 5(2), 1xxx ISSN 1750-1911 1
Review Hunter & McEwen

Table 1. Neuroepigenetic changes induced by stress across the lifespan.


DNA methylation Histone modifications ncRNA
Prenatal stress
GR 17, CRH [66] Altered miRNA to b-glycan [67]
11b-HSD [69]
Reelin, GAD67 [71]
DNMT1, 3A gene expression
Postnatal stress
GR 17 [82] H3K9ac [82]
AVP [101]
BDNF [105]
Adolescent stress

of
? ? ?
Adult acute stress & fear learning
Reelin, PP1 [122] H3Ac [121] , H3S10p-K14Ac [131] Retrotransposon RNA [155]

ro
BDNF [123]
DNMT 3a,b gene expression H3K9me3 miR-18, 24a effect GR [158,159]
[122] H3K27me3 [152]
DNMT inhibition impairs learning H3K4me, H3K27me [153] mir34c [161]
rP
HDAC inhibition improves
learning
Adult chronic stress
DNMT3a gene expression [149] Altered H3K9/K27 methylation miR-186, 709 region-specific
[149] changes [163]
ho

CRH [148] H3K27me3 (social defeat) [143] miR-29bc, 212, 410, 708
chronic sleep deprivation [164]
miR-22, 99a, 137, 2172, let7e
[164]

Aging stress
ut

? ? ?
DNA methylation changes refer to gene promoters or enhancers showing changes in methylcytosine levels, or in
expression of DNA methyltransferases (DNMT). Histone modifications refer to global changes in a particular histone mark
in a part of the brain. The column for ncRNA refers to changes observed in the levels of various ncRNAs such as miRNAs
(e.g.,miR, let) or transcripts from transposable elements.
A

(PTSD) and Type2 diabetes [15] . In PTSD, a enlarge the hippocampus within a time frame
smaller hippocampus, possibly a reflection of of 612 months [6,15] .
low self-esteem [16] , is recognized as both a risk The human brain appears not only receptive
factor for PTSD, as well as a target of chronic to favorable physical stimuli, but also to support-
stress associated with PTSD [17] . There is also a ive relations and meaningful work throughout
smaller hippocampus reported in chronic jet lag life. Structural brain changes have been asso-
[18] , as well as after a 20-year history of chronic ciated with successful cognitivebehavioral
perceived stress, and also with increased levels therapy [19,20] , and it has been suggested that
of systemic inflammation, as well as a lack of psychotherapy can be considered an epigenetic
exercise. The amygdala is more active in anxiety intervention, alongside drug therapy [21] .
disorders and depression, while the prefrontal The health of a persons brain is not only
cortex becomes less efficient with lack of exer- important for effective processing of inputs
cise and also with higher levels of perceived from the external environment, it also controls
stress. Regular moderate exercise has been adjustments of the body engendered by behav-
shown to improve prefrontal cortical blood ioral states like waking, sleeping, lying, standing
flow and function in decision making and also and exercising, and does so via the bidirectional

2 Epigenomics (2013) 5(2) future science group


Stress & anxiety across the lifespan: structural plasticity & epigenetic regulation Review

Development

Stress
St

Histone
modification
DNA
methylation
Control
ncRNA

of
Translation,
mRNA stability Subordinate

Brain Epigenome Brain plasticity Behavior

ro
Figure 1. The brain is the principal perceiver of stress, and it in turn encodes stressful
experience epigenetically. Epigenetic changes alter brain function and brain plasticity, these in
turn alter behavior and cognition. The developmental period affects the brainenvironment
interaction at all levels.
rP
Adapted from [202] .

communication between the neuroendocrine, stress of the mother can impair features of nor-
autonomic, immune and metabolic systems and mal brain development [26] , and that prolonged
the brain [10,22] . These adjustments in the body separation of infant from mother also impairs
promote adaptive activities, such as locomotion, other aspects of brain development and function
ho

and promote coping with aversive situations and [2729] . On the positive side, good maternal care
discrete stimuli, such as noise, crowding, hunger, and consistency of that care is a powerful deter-
excessive heat or cold, and other threats to the minant of life-long patterns of reduced anxiety
persons integrity and safety. Therefore, changes and efficient stress reactivity, as well as social
in the neural architecture of key brain regions and cognitive development [3032] . Moreover, in
ut

will affect how the person handles daily experi- rodents, there are transgenerational effects that
ences, so as to either increase or moderate how a appear to be behaviorally transmitted by the
variety of experiences affect the rest of the body mother to the female offspring [33] , and involve
and ultimately, the fate of the individual. The epigenetic modifications of glucocorticoid recep-
A

foundations of these interactions are established tors, among other genes [34] . In contrast, incon-
early in life. sistent maternal care and maternal anxiety, for
While the focus of this review is upon the example, from food insecurity, produce anxiety
interaction of developmental history with stress, in offspring and appear to contribute to metabolic
brain plasticity and epigenetics, it is important to syndrome and predisposition to diabetes, which
note that biological sex has a significant influence itself has adverse effects on the brain [35,36] . Thus,
on all of these phenomena. Epigenetic factors, the behavioral and physiological consequences
in turn, play a role in the development of sex- of early-life abuse and neglect are profound, and
specific brain structural and functional dimor- the epigenetic concept of behavioral transmission
phism [2325] . This is of particular importance of abuse and its effects on human brain func-
to our translational understanding of these sub- tion are being explored at the level of epigenetic
jects, given the substantial sex bias observed in a regulation of gene expression[37] .
number of brain disorders from autism to anxiety During recent decades, a number of studies
disorders. have documented that early-life adversity also
becomes bodily embedded in human fetuses
Early-life experiences become and children, with major implications for health
biologically embedded throughout the lifespan (for example, see refer-
Animal models have taught us that prenatal ences[38,39]). Chronic dysregulation of adaptive

future science group www.futuremedicine.com 3


Review Hunter & McEwen

systems (allostatic overload), involving epigen- the stress axis and brain plasticity. For example,
etic mechanisms, shapes the developing brain more intense interventions, such as maternal
and bodys biological vulnerability to disease, as separation (MS) have a sensitizing impact on
well as its responsiveness to potential interven- the stress axis, while less severe interventions,
tions [22,40] . Of particular relevance for children like neonatal handling or maternal exercise, can
are experiences of abuse and neglect [41] . On the promote increased resilience to stress in later life
physiological level, adverse childhood experi- [52,53] .
ences are associated with dysregulated cardiovas-
cular, metabolic and immunological function, Prenatal stress
which in turn feed into numerous systemic and Stress and corticosteroid exposure during pre-
behavioral disease conditions [41] . Chaos in the natal life also have the capacity to alter stress
home and inconsistent parenting impairs brain responses, behavior and brain function in later
development, and this can lead to disturbed cog- life [1,54,55] . Prenatal stress or glucocorticoid
nitive function, instable mood, low self-esteem treatment produce lasting behavioral changes
and numerous unhealthy activities, including across the lifespan, including learning deficits

of
overeating, substance abuse, sexual acting-out and increased depressive and anxious behavior
and other forms of legal or illegal risk-taking [42] . [56,57] . These effects are mediated in part by
For example, a 10-year history of a child grow- their effects on stress-responsive brain regions
ing up in a home where the mother has chronic like the hippocampus, amygdala and prefron-

ro
depression is associated with the child having tal cortex, and include reduced dendritic spine
a larger amygdala [43] . Furthermore, low self- density and lower levels of hippocampal neu-
esteem and locus of control is associated with rogenesis [5861] . As with stressors at other life
a smaller hippocampus and a lesser ability to stages, stress intensity influences the outcome
turn off the cortisol stress response [16] . Children for the animal in later life. More severe stress-
rP
growing up without adequate verbal stimulation ors produce exaggerated stress-evoked release of
and in chaotic, unstable home environments corticosterone and impairments in hippocampal
are vulnerable to impaired cognitive function, function [6163] , while milder exposures produce
increased systemic inflammation, cardiovascular improved resilience [64,65] .
disease, substance abuse, antisocial behavior and A number of recent studies have shown that
ho

depression [4447] . prenatal stress results in lasting effects on the


epigenome in a number of stress-sensitive brain
Neuroepigenetic effects of stress in regions. It has been recently demonstrated that
early life prenatal stress causes hypermethylation of the
It is well-established that exposure to environ- GR 17 promoter in the hippocampus and
ut

mental stressors in early life has lasting effects on hypomethylation of the CRH promoter in the
stress reactivity and hippocampal plasticity that hypothalamus and central amygdala of adult
persist into adulthood. In humans, separation male mice, but not females. These differential
from both parents during early life can have a responses correlate with the expression of a
A

decades-long influence on hypothalamicpitui- number of genes in the placenta, including the


taryadrenal (HPA) axis reactivity and liability DNA methyltransferases DNMT1 [66] ; although
to depression. Infants whose mothers suffered these findings remain descriptive, they outline
from starvation during the Dutch famine dur- an interesting area for continued research. Some
ing World WarII had higher levels of depression of the effects observed in this model were passed
and schizophrenia, higher HPA axis reactivity onto the F2generation and were found to result
and twice the rate of cardiovascular disease from altered expression of miRNAs targeting
than the children of properly nourished moth- the b-glycan gene [67] . The effects observed by
ers [48] . These children also had reduced DNA the Bale laboratory [66,67] resemble the effects
methylation of the IGF2 gene 60years later [49] . of maternal depression on GR methylation in
More compelling, from the epigenetic perspec- human infants [68] . Prenatal stress also has the
tive, are the findings that vulnerability to PTSD capacity to change the expression and DNA
and altered HPA axis reactivity can be passed methylation status of the corticosteroid meta-
maternally to the children of Holocaust survi- bolic enzyme 11b-HSD2, and cause increased
vors [50,51] . As these results suggest, severe stress- DNMT3a expression in placenta and increased
ors in early life can have pronounced epigenetic DNMT1 expression in the fetal cortex [69] . In
effects on the stress axis; however, different stress humans, increased 11b-HSD2 methylation is
modalities can have opposing effects on both associated with slower newborn growth and

4 Epigenomics (2013) 5(2) future science group


Stress & anxiety across the lifespan: structural plasticity & epigenetic regulation Review
aberrant behavior [70] . In mice whose dams mothers had higher layers of DNA methylation
were subjected to restraint stress during gesta- at the 17 promoter of the GR in their hip-
tion, increased expression of DNMT1 and 3a pocampus than high LG-ABN offspring, and
and decreased RELN and GAD67 in prefrontal that this change correlated with the behavioral
and hippocampal GABAergic interneurons was changes observed in the model. When pups
seen. The promoters of the reelin and GAD67 from low LG-ABN mothers were cross-fostered
genes also showed increased DNA methylation to high LG-ABN dams, the methylation of the
and hydroxymethylation. These mice also dis- 17 promoter was reduced to levels equivalent
played increased locomotor activity, decreased to those found in the natural offspring of those
social interaction, impaired prepulse inhibi- dams. This demonstrated that the change was
tion and poorer fear conditioning, which were dependent on maternal care rather than genetic
reversed by treatment with valproic acid (a his- or inutero influences. The maternal behavior-
tone deacetylase [HDAC] inhibitor) or clozap- dependent methylation in the 17 promoter
ine (which has DNA demethylase activity) [71] . was most clearly associated with that portion
Both the behavioral and transcriptional changes of the promoter bound by NGFI-A (or egr1).

of
observed in this model are consistent with those Chromatin immunoprecipitation of NGFI-A in
observed in humans suffering from schizophre- the hippocampus of 6-day-old pups showed that
niaa disorder in which maternal stress and binding to the 17 promoter was lower in low
gestational insults have a strong influence [72,73] . LG-ABN pups than in high LG-ABN, although

ro
Neonatal handling can also reduce the long-term total NGFI-A protein levels remained the same
physiological and behavioral consequences of across groups. Low LG-ABN pups also showed
prenatal stress [74] . These results suggest that lower levels of histone acetylation at the 17 pro-
either pharmacological or structured behavioral moter than high LG-ABN. HDAC inhibitors are
interventions in early life (e.g.,[75]) could have capable of both increasing histone acetylation
rP
substantial translational utility in ameliorating levels and reducing levels of DNA methylation
the effects of prenatal stressors. [83] , so Weaver used hippocampal infusions of
the HDAC inhibitor trichostatin A in adult rats
Postnatal stress to successfully reverse both the reduction in hip-
The best-established model for the epigenetic pocampal GR and the increased stress reactivity
ho

impact of early-life adversity is that based on found in low LG-ABN offspring [82,84] . This was
natural variations in maternal care in rodent the first demonstration that a transgenerational
mothers. Michael Meaney and collaborators epigenetic change was reversible with a phar-
have established that rodent mothers, which macological intervention, a finding with obvi-
exhibit high or low levels of arched back nurs- ous implications for human mental disorders
ut

ing (ABN) and maternal licking and grooming associated with childhood abuse or neglect.
of pups (LG), produce offspring with differing The human equivalent of the rodent 17 pro-
stress-related behavior and HPA reactivity in moter is the 1-F promoter [85] , and it too has been
adulthood, mediated in part by higher levels of shown to be hypermethylated in suicides with a
A

hippocampal GR and lower hypothalamic CRH history of childhood abuse [86] ; a more recent
in the offspring of high LG-ABN mothers [7678] . study demonstrated that these effects extended
At the level of synaptic plasticity and learning, to the expression and methylation of the 1-B,
low LG-ABN rearing produces shorter dendritic 1-C and 1-H promoters as well [87] . It should be
arbors and lower spine density in the hippo added that these findings are complex (1-H is
campus, but actually produces improved learn- hypermethylated but the transcript is increased),
ing under stressful conditions [79,80] , a finding and that there are now a small body of stud-
consistent with the adaptive calibration model ies (e.g.,[88]) that suggest simple connections
of stress responsivity [81] . Most remarkably, these between GR methylation status and mental
effects were epigenetic in the strict sense in that disorders that may prove elusive. Childhood
both the variations in stress reactivity and mater- adversity has also been shown to produce hyper-
nal behavior were transmitted across multiple methylation of the GR promoter in leukocytes
generations. from normal adults, and this result was found
The mechanism by which these transgenera- to correlate with a reduced cortisol response to
tional effects occurred was outlined in detail in a dexamethasone CRH challenge [89] . Another
a later paper produced by the Meaney group in recent study using a genome-wide approach
collaboration with the Szyf laboratory [82] . They found that a history of early-life adversity cor-
showed that the adult offspring of low LG-ABN related with differential DNA methylation at

future science group www.futuremedicine.com 5


Review Hunter & McEwen

362 separate promoters, and that this group was due to increased adrenocorticotrophin release in
enriched for genes involved in neuronal plasticity response to stress [100] , which is in turn under the
[90] , further establishing that early-life adversity control of CRH and AVP secreted from the para-
in rodents and man has lasting epigenetic effects ventricular nucleus of the hypothalamus. Mur-
on physiology and behavior. gatroyd etal. examined paraventricular nucleus
While the LG-ABN is the best-known model expression of these two genes in MS animals and
in which transgenerational epigenetic effects on found that MS increased AVP, but not CRH
the stress axis are observed, such effects have expression. The change in AVP expression was
also been described in other model systems. For associated with DNA hypomethylation in an
instance, prenatal treatment with the synthetic enhancer region of the AVP gene bound by the
glucocorticoid, betamethasone, produces sub- methyl CpG-binding domain protein MeCP2
stantial alterations in the global DNA methyla- [101] . MeCP2 is involved in the pathology of
tion status of both fetal organs and the placenta. Rett syndrome [102] , and has been shown to be
Betamethasone also changed the expression involved in the regulation of stress and plasticity
levels of a number of epigenetically significant genes such as BDNF [103,104] . A similar model,

of
genes, notably DNMT3b and Crebbp [91] . Inter- using stressed and abusive dams, caused a reduc-
estingly, some cases of transgenerational epigen- tion in the expression of BDNF in the prefron-
etic effects in animal models appear to be trans- tal cortex of the pups and this reduction was
mitted paternally rather than maternally. The correlated with hypermethylation of the exon

ro
Crews and Skinner laboratories have shown that IV promoter of the BDNF gene. This change
the antiandrogenic fungicide vinclozolin has was found to be reversible when the pups were
transgenerational effects on anxiety and stress- treated with the DNA methylation inhibitor
evoked behavior [92,93] . These alterations seem zebularine [105] . Although not linked to stress
to be transmitted via the paternal germline by perse, differences in anxiety in the high and low
rP
altered DNA methylation of the sperm genome response to novelty rat lines has been linked to
[94] . Another potential case of paternal epigenetic developmental differences in DNMT1 expres-
inheritance has been described by Dietz etal. sion in the hippocampus and amygdala [106] ; as
[95] , who found that the offspring of socially these differences are found largely in the first
defeated fathers were more susceptible to sub- week of life, they are further evidence of the
ho

maximal defeat in adult life than the offspring epigenetic sensitivity of this life stage. The find-
of control fathers. However, invitro fertilization ings reviewed here have firmly established the
did not transmit the increased susceptibility to power of postnatal environment to impact the
defeat. One could conjecture that the effect epigenome, the stress axis, behavior and synaptic
may relate to differential maternal investment plasticity.
ut

based on perceived mate quality, but this has


yet to be established. A neonatal manipulation, Adolescent stress
which shows both maternal and paternal trans- The adolescent life stage marks the transition
mission, is lipopolysaccharide treatment, which from the rapid growth that characterizes infancy
A

alters anxiety behavior and stress-evoked corti- and childhood, towards the utilization of the
costerone to at least the F2generation. Maternal organisms energetic resources for reproductive
transmission was blocked by cross-fostering, and purposes. As such, it marks the point at which
seems to occur via maternal behavior. However, most mammals turn outward from parental
the mechanism of paternal transmission in this attachments towards a wider social milieu. The
model is, as yet, unclear [96] . demands of that milieu are likely more severe in
MS is another model of early-life stress and animals with complex societies, such as human
adversity. MS paradigms typically involve sepa- beings. Adolescence is marked by a number
rating the pups from their dam for several hours of changes in behavior, brain structure and
a day during the first 2weeks of post-natal life. function [107] . It is also the time of life where
Like the LG-ABN model, MS can have trans- humans are most likely to develop a variety of
generational effects on stress reactivity, although psychiatric disorders including substance abuse,
it too appears to act via paternal transmission schizophrenia and depression [108] . Why this
[97] and have a differential effect on males and is the case, when many of these disorders are
females. Much like low LG-ABN rearing, MS associated with stress exposure in earlier life, is
results in increased HPA reactivity and clear unclear. However, one could hypothesize that
effects on hippocampal synaptic plasticity and the increased demand for social cognitive capac-
learning in adulthood [29,98,99] . This is, in part, ity at this age could reveal pre-existing deficits

6 Epigenomics (2013) 5(2) future science group


Stress & anxiety across the lifespan: structural plasticity & epigenetic regulation Review
in stress-sensitive regions, such as the amygdala, methylation might undergird the formation of
which play a role in social learning [109] . memory [118120] , but the involvement of this
It is clear that adolescence is also a transition epigenetic mark in memory was only recently
in the development of the stress axis and upon established, and the first demonstration of an
hippocampal plasticity [1,54,55] . Human adoles- epigenetic modification playing a role in fear
cents have increased basal and stress-evoked conditioning regarded histone acetylation [121] .
HPA activity [110] , and peripubertal rats also Miller and Sweatt demonstrated that fear con-
show a potentiation of adrenocorticotrophin ditioning increases the expression of DNMT3A
and corticosterone in response to stress [111,112] . and 3B in the hippocampus, and that treat-
Most significantly from the perspective of ment with DNA methyltransferase inhibitors
stress-induced plasticity, peripubertal exposure impaired memory consolidation [122] . Fear con-
to chronic variable stress reduces adult hippo ditioning also altered the methylation status of
campal volume and spatial cognition [113] . While the reelin, PP1 and BDNF genes [122,123] . PP1 is
it is readily apparent that adolescence is a sig- of interest as it plays a role as a phosphatase at
nificant neurodevelopmental stage, compara- histone H3 serine10, and this activity would

of
tively little is known about the effects of stress appear to be the basis for its role in memory
on brain structural and functional plasticity at formation [124,125] . Similarly, Maddox and Schafe
this time. Little has been reported with regard have shown that HDAC inhibition in the lateral
to alterations in epigenetic phenomena. Given amygdala enhances fear memory reconsolida-

ro
the importance of the adolescent transition in tion, while inhibition of DNMTs interferes with
the development of human mental disorders, it reconsolidation [126] .
is to be hoped that this dearth of knowledge can A series of studies performed in the Tsai labo-
soon be remedied. ratory established a role for histone acetylation
in memory formation as well. Beginning with
rP
Neuroepigenetic effects of stress in the observation that environmental enrichment
adult life ameliorated memory deficits in a transgenic
The hippocampus, a key brain structure for mouse model of neurodegenerative disease,
episodic and spatial memory and also mood these investigators explored the possibility that
regulation, is a highly stress-sensitive tissue and, these effects might be replicated by treatment
ho

as a result, it has received substantial attention with an HDAC inhibitor, as in fact proved to
from stress, learning and memory and mood be the case as HDAC inhibition improved both
disorder researchers. The hippocampus shows memory and synapse formation in the anterior
plasticity in response to both acute and chronic cingulate cortex [127] . Subsequently, the same
stressors, as well as antidepressant treatments, group showed that these effects were regulated
ut

and also spatial and episodic learning tasks; via the classI HDAC, HDAC2, which appears
indeed, many learning and memory tasks are to be the major HDAC of its class in neurons.
stressful, and many stressors produce some HDAC2 did so by regulating the expression
learning (e.g.,[114]). The adult hippocampus of a relatively small group of genes, including
A

also expresses as large a variety of epigenetic a number of glutamate receptor subunits and
enzymes as any other brain region, as a perusal BDNF [128] , the latter finding replicating an ear-
of the Allen Brain Atlas will demonstrate [115] . lier one by Bredy etal. [129] . A significant linkage
Therefore, the hippocampus has proven to be between epigenetic chromatin modifications and
fertile ground for the study of the intersection synaptic plasticity was recently established by
of stress, plasticity and epigenetics, although, as Calfa and collaborators, who found that inhibi-
we shall see, it is not the only brain region where tion of HDACs blocked the capacity of BDNF
this intersection can be found. to increase both spine density and the volume of
Fear conditioning, which is by definition psy- excitatory neurotransmitter release in the CA1
chologically stressful, has been of great use as of the rat hippocampus [130] .
a behavioral probe of the epigenetic influences One of the earliest findings explicitly
on learning and plasticity in animal models. As implicating stress in the regulation of epigen-
this area has been the subject of recent, system- etic marks in brain was that of Bilang-Bleuel
atic review, both in this journal and elsewhere etal. who showed that an acute stress, forced
[116,117] , we will not attempt a comprehensive swimming, increased the dual phospho-acetyl
treatment here; rather, we will focus on salient mark at serine 10 and lysine 14 of histone H3
findings with relevance to stress-induced plas- (H3S10p-K14ac) in the rat dentate gyrus [131] .
ticity. It has long been hypothesized that DNA The H3S10p-K14ac mark is associated with

future science group www.futuremedicine.com 7


Review Hunter & McEwen

transcriptional activation [132,133] , and previous tri-methylation in the nucleus accumbens also
work had shown it to be associated with treat- change in response to cocaine administration,
ment with a variety of neurotransmitter agonists where this mark is also associated with changes
[134] . Building upon this finding, the Reul group in dendritic spines [151] . In the hippocampus,
showed that the same effect was observable this mark is profoundly upregulated by acute
with a different stressor, and that it was NMDA stress or by chronic treatment with the anti-
receptor-dependent and associated with neuro- depressant fluoxetine [152] ; the same study also
nal activation (as measured by c-fos induction) demonstrated stress-dependent changes in both
in the same cells that showed changes in the the H3K4me3 (moderately increased in chronic
double mark [135,136] . Exercise, which may be stress) and H3K27me3 (substantially reduced by
regarded as a positive stressor [54] , also increases acute stress) marks in the hippocampus. Both
levels of the mark, suggesting that changes in H3K4 and H3K27 methylation were found to
H3S10p-K14ac may be an adaptive, rather than be reduced at the prodynorphin promoter in the
pathogenic, response to stress [137] . GABAergic striatum of rats subjected to an acute swim stress
interneurons appear to be a countervailing force [153] . The balance of evidence from these studies

of
on stress-induced increases in H3S10p-K14ac suggests that interventions that increase levels of
[138] . Given the established synergy between the the H3K9me3 mark or inhibit HDAC2 can pro-
glutamate and glucocorticoid systems in produc- mote resilience to stress and depressive behavior
ing stress-induced plasticity [139] , it is of interest in animal models and, by extension, also in man.

ro
that stress-induced increases in H3S10p-K14ac Our finding that acute restraint stress rapidly
require both systems acting through the novel and globally upregulates the H3K9me3 mark in
intermediaries Elk-1 and MSK1. The GR inter- a hippocampus-specific fashion [152] , led us to
acts directly with these proteins, and this inter- hypothesize that this mark might be involved
action appears to promote their phosphorylation in suppressing the expression of transposable
rP
by the ERK-MAPK pathway acting downstream elements, which can be induced by stress [154] .
of activated NMDA receptors. Furthermore, Tight control of genomic stability in terminally
this coinduction of the mark plays a role in the differentiated neurons is of obvious importance,
memory of the stress that produced it [140,141] . and so we used chromatin immunoprecipitation
Social defeat stress, which is one of the better sequencing of the H3K9me3 mark to deter-
ho

rodent models of human depression in terms of mine if in fact this mark showed a selective bias
ethological and face validity [142] , also has an towards accumulation at transposable elements
impact on the neural epigenome, as was first after acute stress. Our results showed that, while
demonstrated by the Nestler laboratory [143] . there was little directionality to the changes in
They showed that chronic social defeat in adult H3K9me3 levels on genes, stress produced an
ut

mice significantly increased the levels of the increase in the mark at more than 50-fold more
repressive H3K27 trimethyl mark at the BDNF transposable elements than elements that saw a
promoter, while treatment with antidepressant decrease, accounting for at least 50,000 distinct
drugs increased marks associated with active loci. This effect was accompanied by a 50% or
A

transcription, such as H3 acetylation and H3K4 more decrease in the expression of retrotranspo-
methylation [143,144] . Later studies by the same son RNAs, notably the B2 SINE and IAP endog-
group linked similar changes with changes in enous retrovirus (see Figure2 ) [155] . These results
HDAC function in the nucleus accumbens after point to an entirely novel area of investigation
stress or cocaine treatment. Notably, they found in the neurobiology of the stress response, sug-
that HDAC2 had an antidepressant effect in the gesting that a large and hitherto underexplored
defeat model [145,146] . A further link with the portion of the genome may be involved both in
epigenetic actors in learning and memory was the normal response to stress and, potentially,
the discovery that defeat increased accumbal in the pathogenesis of stress-related disorders.
DNMT3A expression, while chronic cocaine The latter possibility has been given more weight
reduced it, and that these changes were associ- by the recent finding that LINE-1 and Alu ret-
ated with plastic changes in dendritic spines in rotransposons were differentially DNA methyl-
the same brain region [147] . Resilience to social ated between veterans with PTSD and combat-
stress also correlates with altered DNA meth- deployed controls [156] . Transposable elements
ylation of the CRH gene in the paraventricular can of course contribute to genomic instability,
hypothalamus [148] and levels of the repressive but they may also have significant roles in the
H3K9 and K27 trimethylations in the accum- regulation of transcription, RNA stability and
bens [149,150] . Levels of histone H3K9 di- and so on [157] .

8 Epigenomics (2013) 5(2) future science group


Stress & anxiety across the lifespan: structural plasticity & epigenetic regulation Review
Relative to DNA methylation and chromatin expression of the CRHR1 receptor and, thereby,
modifications, ncRNAs have received less atten- reduces anxiety [161] . In the mouse frontal cortex,
tion with regard to stress and brain plasticity. acute stress selectively alters the expression of
Nonetheless, a number of miRNAs have been Let-7a, miR-9 and 26a/b, while it has no effect
identified that are involved in the regulation of on these miRNAs in the hippocampus [162] . Sim-
GR, including miR-124a and miR-18a [158,159] . ilarly, regionally specific effects on miR-186 and
Both acute and chronic stress regulate the expres- 709 were found in the hippocampus, prefrontal
sion of miR-134 and -183 in the hippocampus cortex and cerebellum of chronically stressed rats
and amygdala, and through them alters the [163] . Sleep deprivation stress altered the expres-
splicing of acetylcholinesterase mRNA, poten- sion of ten miRNAs in mouse brain, seven of
tially allowing for fine-tuning of the cholinergic which appear to be corticosteroid regulated
responsiveness of these regions [160] . Acute and [164] . While all of the findings reviewed above
chronic stress also modulate the expression of involve miRNA, other species of ncRNA are
miR-34 in the amygdala, where it suppresses the likely to be involved in the brains adaptation or

of
20,000

15,000
Number of islands

ro
Stress
Stress up
10,000 Stress down
rP
5000

0
ERV/LTR

Satellite
LINE

SINE

DNA

ncRNA
Simple/low
complexity/
other

ho

Hippocampus RNA
2
ut

0 Genomic Transcriptional
Fold change

instability? stability?
-2
A

-4 *

**
-6
B2 control

B2 stress

IAP control

IAP stress

L1 control

L1 stress

Figure 2. Epigenetic regulation of mobile genomic elements. (A) Displays a comparison of


transposable element loci, broken down by taxon, that showed either an increase in H3K9me3 levels
after stress (stress up) or a decrease (stress down). Retrotransposons (LINEs; ERV/TR; SINEs) and
simple repeats were enriched 50-fold or more, while satellite repeats, DNA transposons and ncRNAs
were less enriched. (B) Shows that this increase was accompanied by a hippocampus-specific
suppression of the expression of RNAs for several of the elements. Other tissues showed no such
change. These findings raise the possibility that loss of epigenetic control could contribute to
transposon-induced genomic and transcriptional instability, contributing to brain weathering.
ERV: Endogenous retrovirus; IAP: Intracisternal A particle; LINE: Long interspersed nuclear elements;
LTR: Long terminal repeat; SINE: Short interspersed nuclear element.
Adapted from [155] .

future science group www.futuremedicine.com 9


Review Hunter & McEwen

maladaptation to stress. The recent finding that involved in the aging process is only beginning
piRNAs, whose actions were long thought to be to be understood.
restricted to the gonads, play an active role in It is evident that aging is associated with
synaptic plasticity in the Aplysia [165] , raises the changes in the epigenome; indeed, a global com-
question as to their involvement in mammalian parison of methylated DNA between human
synaptic plasticity, memory and stress. Similarly, newborns, young adults and centenarians found
in Arabidopsis thalania, transposable element that extreme old age was associated with a sub-
RNA has the capacity to regulate the plants stantial decline in overall DNA methylation in
stress response via the siRNA pathway [166] . Our lymphocytes [177] . In the brain, however, DNA
own observations that the RNAs for a number of methylation increases with age, as is evident in
transposable elements are regulated by stress in some animal models and human studies [178181] .
the hippocampus, suggests that there may be a Furthermore, evidence is emerging that some dis-
role for these ncRNAs in the mammalian stress orders of aging, such as Alzheimers disease, may
response as well. be associated with even greater increases in levels
of methylated DNA in the brain [182] . DNMT3a

of
Neuroepigenetic effects of stress in expression in the hippocampus increases with
aging age and, interestingly, caloric restriction, which
Aging is associated with changes in the stress is thought to promote healthy aging [183] , reduces
axis, cognition and neuronal plasticity, and this increase [184] . As caloric restriction tends to

ro
is regarded by some as the last epigenetically increase circulating corticosteroids [185] , this study
regulated stage of development [167,168] . While raises interesting questions about how a life his-
aging is generally viewed as a gradual process, tory of stress might alter the DNA methylation
it includes more clearly demarcated endocrine profile in the aging brain, although it is worth not-
events relevant to both stress responsivity and ing that corticosteroids can have positive effects
rP
brain plasticity, such as menopause and adreno- on plasticity and that stress-mediated changes
pause. However, the degree to which these events involve other molecules, such as glutamate [186] .
might be under epigenetic control is not yet On the other hand, another recent study found
clear. In the aged rat, the HPA axis response to reduced levels of DNMT3a in aged mouse brain
stress is prolonged when compared with young and showed that increasing the levels of the 3a2
ho

adult animals, and GR levels decline [169,170] . version improves cognition. This is an issue that
These findings by Robert Sapolsky led him to will have to be resolved with further experimenta-
propose the glucocorticoid cascade hypothesis tion, with particular attention paid to the stress
of aging in 1986 [171] , which proposes that age- history of the animals. As has been well estab-
induced decline in GR feedback exposes the lished, here and elsewhere, different stress his-
ut

brain to progressively higher levels of cortico- tories can produce widely divergent outcomes in
steroids, which, in turn, impairs hippocampus brain and behavior. Brain DNA hydroxymethyl-
structure and function and leads to cognitive ation also appears to increase with age and due to
decline. While this hypothesis has been modi- its poorer interaction with DNA methyl-binding
A

fied to de-emphasize neuronal loss and empha- proteins is likely to interfere with their function
size age-related loss of structural and functional [187,188] , and, by extension, cognition.
plasticity [172] , it is evident that stress and HPA Several groups have found that aging and
activity have a role in vulnerability to brain aging neurodegeneration is associated with increased
in both humans and animals [1,173] . As is readily HDAC2 expression in the hippocampus and
apparent, aging is often associated with cognitive reduced acetylation at the H3K9 and H4K12
decline, and some of this decline with reduced loci, and that age-related cognitive deficits could
plasticity. In plastic, stress-sensitive regions, such be reversed with enhanced BDNF signaling,
as the hippocampus, aging is associated with HDAC inhibitors or shRNA directed against
reductions in synapse number, neurogenesis HDAC2 [189191] . Most salient in terms of the
and long-term potentiation, accompanied by an present discussion is the finding that the increase
increase in long-term depression after an acute in HDAC2 expression in the neurodegenerat-
stressor [174176] . In the aged rat prefrontal cortex, ing brain is driven by the binding of GR to the
chronic stress retains the capacity to reduce den- HDAC promoter, clearly linking the stress axis
dritic complexity, but the region appears to have with what is, at present, the best understood epi-
lost the resilience present in young adults and genetic mediator of age-related cognitive decline
does not recover to prestress levels of structural [189] . In contradistinction to these findings, other
complexity [12] . How stress and epigenetics are groups have found that histone H4 acetylation

10 Epigenomics (2013) 5(2) future science group


Stress & anxiety across the lifespan: structural plasticity & epigenetic regulation Review
and H3K9 trimethylation increase with age in foremost in the minds of neuroepigeneticists, and
mouse hippocampal and cortical neurons, and as a result a certain amount of confusion in the
these increases are greater in neurons from the literature is likely to occur. We would suggest that
3xTg transgenic model of Alzheimers disease [192] , controlling for stress is of substantial importance
and the increase in H3K9me3 was associated with for the fields rapid advancement.
decreased BDNF expression. Of course, a global Future work in the area will have to go further
increase in histone H4 acetylation is likely sepa- in describing the mechanisms by which epigenetic
rable from a decrease in the acetylation of one of changes persist and explain their regional selectiv-
many basic amino acid residues in the H4 tail. ity. In the latter case, increasing the level of ana-
Indeed, there is some evidence that the true pic- tomical detail involved in neuroepigenetic analysis
ture may be more complex than either of these will be crucial. Technical advances in next-gener-
narratives would suggest. Castellano recently ation sequencing will drive some of this, as will
demonstrated that the subregional hippocampal techniques for the analysis of specific cell types,
epigenetic profile of spatial learning in age unim- such as bacterial artificial chromosome-translating
paired rats is largely indistinguishable from young ribosome affinity purification [194,195] or circuit-

of
rats with the exception of an increase in global based analyses using optogenetics, as will more
H4 acetylation, which was associated with age in established approaches.
both impaired and unimpaired animals. What As we have pointed out above, the adolescent
differed in age-impaired animals was an inabil- period, in spite of its obvious importance as a

ro
ity to reduce HDAC2 expression in response to period of increased susceptibility to a variety of
behavioral activation [193] , and a lack of coor- stress-related mental disorders [196], has had little to
dination of epigenetic changes with memory no examination of the impact of stress on epigen-
performance. Another important finding of the etic marks in the brain. Nor has much attention
latter study was that several commercial antibod- been paid, as yet, to how stress-induced epigenetic
rP
ies against specific histone modifications lacked changes at one life stage, such as infancy, might
the advertised specificity, which is worth noting, interact with stress, and susceptibility to stress in
both in evaluating the literature to date, and in later life stages, such as adolescence or aging.
designing future studies in the field of neuroepi- Much of the research to date on the neuro
genetics. While we now have the basic lineaments epigenetics of stress has logically focused on well-
ho

of an understanding of some of the interactions known genes, for example, GR and BDNF. It is
between the stress axis and epigenetic and syn- likely, however, that these are not the only genomic
aptic plasticity in the brain, it should be evident elements involved in the epigenetics of stress and
that much further work is to be done before our anxiety. As an example, the mineralocorticoid
understanding of the impact of stress during this receptor, which plays a significant role in HPA axis
ut

final life stage is fully understood. feedback and anxiety [197] , has yet to be examined
for epigenetic activity in the brain, although it has
Conclusion been associated with altered histone methylation
While the interactions between external stressors, in the kidney [198] . The availability of cheaper and
A

epigenetics and brain plasticity across the lifes- deeper next-generation sequencing technologies
pan have only begun to be explored, it is read- will be of great help in broadening the number of
ily apparent that this area of exploration is offer- potential genomic elements examined to encom-
ing insights into how the environment produces pass more than the genes themselves to the other
persistent changes in the brain and behavior. Of 95% of the genome, most of which may be actively
greater interest from the translational perspective transcribed [199] , although their functions remain
is the fact that many of these changes appear to be largely unexamined.
reversible via structured environmental interven- For basic discoveries about the epigenetics
tions or pharmacologic means. of stress and anxiety to leave the bench and be
A problem for the study of epigenetics in the translated to the bedside, epigenetic pharmacol-
brain is the transient nature of many of the phe- ogy must advance substantially. While many his-
nomena coupled with the necessity for making tone deacetylase inhibitors have been approved for
exvivo observations. As we have argued through- human use, drugs to inhibit or increase histone
out, stress has significant effects on the epigenome, methylation are relatively few and far between,
and thus attention to stress history, reducing the although some methyltransferase inhibitors are
stress of sacrificing experimental animals and rapid becoming available for research purposes [200,201] .
tissue dissection are very important considerations. As to the many other histone modifications or
It is not evident that these concerns are always ncRNA, still less is known.

future science group www.futuremedicine.com 11


Review Hunter & McEwen

Future perspective hoped that as our fundamental understanding of


The field of the epigenetics of stress and anxi- chromatin structure, transcription and the vast
ety has many open questions outstanding. terra incognita of what used to be called Junk
Notably, how epigenetic activity integrates with DNA improves, so will our understanding of
better described systems such as neurotransmit- the specificity of neuroepigenomic phenomena.
ters. Another question is how stable epigenetic Epigenetic mechanisms are an extremely
changes are maintained, as recent research has promising means by which to understand how
revealed that many, if not most, epigenetic marks the slings and arrows of both ordinary and
are much less stable and far more dynamic than extraordinary misfortune affect how our brain
had been thought. One of the greatest open functions across our lifespan, and how some indi-
questions is the issue of specificity in neuro- viduals and certain times of life might be more
epigenetics. This is significant at a number of vulnerable to the development of particular brain
levels. At the level of experimental tools there disorders. As our technical and observational
are few truly specific pharmacophores and capacity improves, it seems likely that we will
genetic manipulations of epigenetic informa- gain a much more profound understanding of

of
tion; although quite useful when well employed, how stress in our environment produces lasting
they can substantially muddy our understanding changes in our brains, mood and behavior.
when they are not. The question of how a rela-
tively nonspecific HDAC inhibitor, for example, Financial & competing interests disclosure

ro
alters the expression of only a small fraction of RG Hunter and BS McEwen thank NIH MH41256 and
genes in a small number of brain regions, as has the Help for Depression Research Foundation (HDRF) for
been observed by a number of groups, is also their support. The authors have no other relevant affilia-
highly significant. One could imagine that the tions or financial involvement with any organization or
combination of chromatin status, and the bind-
rP
entity with a financial interest in or financial conflict with
ing of various elements of the transcriptional the subject matter or materials discussed in the manuscript
machinery, could regulate the accessibility of apart from those disclosed.
certain genes to manipulation, but this hypoth- No writing assistance was utilized in the production of
esis has not yet been tested in the brain. It is to be this manuscript.
ho

Executive summary
Background
Stress has a significant effect on the health of a variety of bodily systems.
ut

Stress effects brain plasticity at a variety of levels.


Stress, particularly chronic and early-life stress, can have long-lasting effects on the brain and behavior.
Sex has a profound influence on stress responsivity and brain plasticity.
Neuroepigenetic effects of stress in early life
A

Early-life stress is well established to have lasting impacts on health in both humans and animal models.
The effects of stressful life experience can be transmitted epigenetically from parents to offspring across multiple generations.
Both pre- and post-natal stress can alter the expression of a variety of epigenetic marks in the brain, with significant consequences for
gene expression and brain function.
Adolescence is one of the most poorly understood periods with regard to the effects of stress on brain plasticity, yet it is likely to be
one of the most important to our understanding of human mental disorders.
Neuroepigenetic effects of stress in adult life
Stress in adult life is well known to influence health and disease.
The brain, particularly regions such as the hippocampus, is sensitive to stress in adult life, as in early life, although the impacts can be
different.
Adult stressors and stressful learning paradigms have been shown to alter a number of different epigenetic pathways in the brain.
Recent studies suggest that stress interacting with epigenetic mechanisms may alter the behavior of transposable elements in the
genome of the hippocampus.
Neuroepigenetic effects of stress in aging
The aging brain is less resilient to stress than during earlier life stages; thus, stress can have a more persistent negative impact on brain
structure and function during aging.
There are epigenetic correlates of aging in the brain, and some of these have shown alteration with stress.

12 Epigenomics (2013) 5(2) future science group


Stress & anxiety across the lifespan: structural plasticity & epigenetic regulation Review
14 Roozendaal B, McEwen BS, Chattarji S. in adult male rat offspring.
References Stress, memory and the amygdala. Nat. Rev. Neuropsychopharmacology 30, 21922204
Papers of special note have been highlighted as:
n of interest
Neurosci. 10, 423433 (2009). (2005).
nn of considerable interest 15 McEwen BS, Gianaros PJ. Stress- and 29 Eiland L, McEwen BS. Early life stress followed
allostasis-induced brain plasticity. Annu. Rev. by subsequent adult chronic stress potentiates
1 Lupien SJ, McEwen BS, Gunnar MR,
Med. 62, 431445 (2011). anxiety and blunts hippocampal structural
HeimC. Effects of stress throughout the
16 Pruessner JC, Baldwin MW, Dedovic K etal. remodeling. Hippocampus 22(1), 8291 (2012).
lifespan on the brain, behaviour and
cognition. Nat. Rev. Neurosci. 10(6), 434445 Self-esteem, locus of control, hippocampal 30 Caldji C, Diorio J, Meaney MJ. Variations in
(2009). volume, and cortisol regulation in young and maternal care in infancy regulate the
old adulthood. NeuroImage 28, 815826 development of stress reactivity. Biol. Psychiat.
2 McEwen BS, Gianaros PJ. Central role of the
(2005). 48, 11641174 (2000).
brain in stress and adaptation: links to
socioeconomic status, health, and disease. 17 Gurvits TV, Shenton ME, Hokama H etal. 31 Akers KG, Yang Z, Delvecchio DP etal. Social
Ann. NY Acad. Sci. 1186, 190222 (2010). Magnetic resonance imaging study of competitiveness and plasticity of
hippocampal volume in chronic, combat- neuroendocrine function in old age: influence
3 McEwen BS, Eiland L, Hunter RG,
related posttraumatic stress disorder. Biol. of neonatal novelty exposure and maternal care
MillerMM. Stress and anxiety: structural
Psychiatry 40, 10911099 (1996). reliability. PLoS ONE 3(7), e2840 (2008).
plasticity and epigenetic regulation as a
consequence of stress. Neuropharmacology 18 Cho K. Chronic jet lag produces temporal 32 Tang AC, Reeb-Sutherland BC, Yang Z,

of
62(1), 312 (2012). lobe atrophy and spatial cognitive deficits. Romeo RD, McEwen BS. Neonatal novelty-
Nat. Neurosci. 4, 567568 (2001). induced persistent enhancement in offspring
4 McEwen BS. Stress, sex, and neural
19 Davidson RJ, McEwen BS. Social influences spatial memory and the modulatory role of
adaptation to a changing environment:
on neuroplasticity: stress and interventions to maternal self-stress regulation. J.Neurosci.
mechanisms of neuronal remodeling. Ann. NY

ro
promote well-being. Nat. Neurosci. 15(5), 31(14), 53485352 (2011).
Acad. Sci. 1204(Suppl.), E38E59 (2010).
689695 (2012). 33 Francis D, Diorio J, Liu D, Meaney MJ.
5 Bennett E, Diamond M, Krech D,
20 Holzel BK, Carmody J, Evans KC etal. Stress Nongenomic transmission across generations of
Rosenzweig M. Chemical and anatomical
reduction correlates with structural changes maternal behavior and stress responses in the
plasticity of brain. Science 146, 610619
in the amygdala. Soc. Cogn. Affect. Neurosci. rat. Science 286, 11551158 (1999).
rP
(1964).
5(1), 1117 (2009). 34 Weaver ICG, Cervoni N, Champagne FA etal.
6 Erickson KI, Voss MW, Prakash RS etal.
21 Stahl SM. Psychotherapy as an epigenetic Epigenetic programming by maternal behavior.
Exercise training increases size of
drug: psychiatric therapeutics target Nat. Neurosci. 7, 847854 (2004).
hippocampus and improves memory. Proc.
Natl Acad. Sci. USA 108(7), 30173022 symptoms linked to malfunctioning brain 35 Kaufman D, Smith ELP, Gohil BC etal. Early
(2011). circuits with psychotherapy as well as with appearance of the metabolic syndrome in
drugs. J.Clin. Pharm. Ther. 37(3), 249253 socially reared bonnet macaques. J.Clin.
ho

7 Wang X, Merzenich MM, Sameshima K,


(2012). Endocrinol. Metab. 90, 404408 (2005).
Jenkins WM. Remodelling of hand
representation in adult cortex determined by 22 McEwen BS. Protective and damaging effects 36 Kaufman D, Banerji MA, Shorman I etal.
timing of tactile stimulation. Nature 378, of stress mediators. N.Engl. J.Med. 338, Early-life stress and the development of obesity
7175 (1995). 171179 (1998). and insulin resistance in juvenile bonnet
23 Nugent BM, McCarthy MM. Epigenetic macaques. Diabetes 56, 15 (2007).
8 Draganski B, Gaser C, Kempermann G etal.
ut

Temporal and spatial dynamics of brain underpinnings of developmental sex 37 Mcgowan PO, Sasaki A, Dalessio AC etal.
structure changes during extensive learning. differences in the brain. Neuroendocrinology Epigenetic regulation of the glucocorticoid
J.Neurosci. 26(23), 63146317 (2006). 93(3), 150158 (2011). receptor in human brain associates with
24 Hoffmann A, Spengler D. The lasting legacy childhood abuse. Nat. Neurosci. 12, 241243
9 Bezzola L, Merillat S, Gaser C, Jancke L.
(2009).
A

Training-induced neural plasticity in golf of social stress on the epigenome of the


novices. J.Neurosci. 31(35), 1244412448 hypothalamicpituitaryadrenal axis. 38 Danese A, McEwen BS. Adverse childhood
(2011). Epigenomics 4(4), 431444 (2012). experiences, allostasis, allostatic load, and age-
25 Bale TL. Sex differences in prenatal related disease. Physiol. Behav. 106(1), 2939
10 McEwen BS. Physiology and neurobiology of
epigenetic programming of stress pathways. (2012).
stress and adaptation: central role of the
brain. Physiol. Rev. 87, 873904 (2007). Stress 14(4), 348356 (2011). 39 Entringer S, Epel ES, Kumsta R etal. Stress
26 Maccari S, Morley-Fletcher S. Effects of exposure in intrauterine life is associated with
11 McEwen BS. Stress and hippocampal
prenatal restraint stress on the hypothalamus shorter telomere length in young adulthood.
plasticity. Annu. Rev. Neurosci. 22, 105122
pituitaryadrenal axis and related behavioural Proc. Natl Acad. Sci. USA 108(33), E513E518
(1999).
and neurobiological alterations. (2011).
12 Bloss EB, Janssen WG, McEwen BS,
Psychoneuroendocrinology 32, S10S15 (2007). 40 Shonkoff JP, Boyce WT, McEwen BS.
Morrison JH. Interactive effects of stress and
27 Francis DD, Diorio J, Plotsky PM, Meaney Neuroscience, molecular biology, and the
aging on structural plasticity in the prefrontal
MJ. Environmental enrichment reverses the childhood roots of health disparities. JAMA
cortex. J.Neurosci. 30(19), 67266731
effects of maternal separation on stress 301, 22522259 (2009).
(2010).
reactivity. J.Neurosci. 22, 78407843 (2002). 41 Anda RF, Butchart A, Felitti VJ, Brown DW.
13 Vyas A, Mitra R, Rao BSS, Chattarji S.
28 Plotsky PM, Thrivikraman KV, Nemeroff Building a framework for global surveillance of
Chronic stress induces contrasting patterns of
CB, Caldji C, Sharma S, Meaney MJ. Long- the public health implications of adverse
dendritic remodeling in hippocampal and
term consequences of neonatal rearing on childhood experiences. Am. J.Prev. Med.
amygdaloid neurons. J.Neurosci. 22,
central corticotropin-releasing factor systems 39(1), 9398 (2010).
68106818 (2002).

future science group www.futuremedicine.com 13


Review Hunter & McEwen

42 Evans GW, Gonnella C, Marcynyszyn LA, age-related glucocorticoid secretion and gene (NR3C1) and infant cortisol stress
Gentile L, Salpekar N. The role of chaos in cognitive performance: a longitudinal study responses. Epigenetics 3(2), 97106 (2008).
poverty and childrens socioemotional in the rat. Eur. J.Neurosci. 11(8), 29062916 69 Jensen Pena C, Monk C, Champagne FA.
adjustment. Psychol. Sci. 16, 560565 (2004). (1999). Epigenetic effects of prenatal stress on 11beta-
43 Lupien SJ, Parent S, Evans AC etal. Larger 57 Vallee M, Mayo W, Dellu F, Le Moal M, hydroxysteroid dehydrogenase-2 in the
amygdala but no change in hippocampal Simon H, Maccari S. Prenatal stress induces placenta and fetal brain. PLoS ONE 7(6),
volume in 10-year-old children exposed to high anxiety and postnatal handling induces e39791 (2012).
maternal depressive symptomatology since low anxiety in adult offspring: correlation 70 Marsit CJ, Maccani MA, Padbury JF,
birth. Proc. Natl Acad. Sci. USA 108(34), with stress-induced corticosterone secretion. LesterBM. Placental 11-beta hydroxysteroid
1432414329 (2011). J.Neurosci. 17(7), 26262636 (1997). dehydrogenase methylation is associated with
44 Farah MJ, Shera DM, Savage JH etal. 58 Seckl JR. Glucocorticoids, developmental newborn growth and a measure of
Childhood poverty: Specific associations with programming and the risk of affective neurobehavioral outcome. PLoS ONE 7(3),
neurocognitive development. Brain Res. 1110, dysfunction. Prog. Brain Res. 167, 1734 e33794 (2012).
166174 (2006). (2008). 71 Matrisciano F, Tueting P, Dalal I etal.
45 Danese A, McEwen BS. Adverse childhood 59 Cratty MS, Ward HE, Johnson EA, Azzaro Epigenetic modifications of GABAergic
experiences, allostasis, allostatic load, and AJ, Birkle DL. Prenatal stress increases interneurons are associated with the
age-related disease. Physiol. Behav. 106(1), corticotropin-releasing factor (CRF) content schizophrenia-like phenotype induced by

of
2939 (2011). and release in rat amygdala minces. Brain Res. prenatal stress in mice. Neuropharmacology
46 Hart B, Risley TR. Meaningful Differences in 675(12), 297302 (1995). doi:http://dx.doi.org/10.1016/j.
the Everyday Experience of Young American 60 Murmu MS, Salomon S, Biala Y, Weinstock bbr.2011.03.031 (2012) (Epub ahead of
Children. Brookes Publishing Company, MD, M, Braun K, Bock J. Changes of spine density print).

ro
USA (1995). and dendritic complexity in the prefrontal 72 Brown AS. The environment and
47 Pesonen AK, Raikkonen K. The lifespan cortex in offspring of mothers exposed to susceptibility to schizophrenia. Prog.
consequences of early life stress. Physiol. stress during pregnancy. Eur. J.Neurosci. Neurobiol. 93(1), 2358 (2011).
Behav. 106(5), 722727 (2012). 24(5), 14771487 (2006). 73 Koenig JI, Kirkpatrick B, Lee P.
48 Roseboom TJ, Painter RC, van Abeelen AF, 61 Lemaire V, Koehl M, Le Moal M, Abrous Glucocorticoid hormones and early brain
rP
Veenendaal MV, De Rooij SR. Hungry in the DN. Prenatal stress produces learning deficits development in schizophrenia.
womb: what are the consequences? Lessons associated with an inhibition of neurogenesis Neuropsychopharmacology 27(2), 309318
from the Dutch famine. Maturitas 70(2), in the hippocampus. Proc. Natl Acad. Sci. (2002).
141145 (2011). USA 97(20), 1103211037 (2000). 74 Lemaire V, Lamarque S, Le Moal M, Piazza
49 Heijmans BT, Tobi EW, Stein AD etal. 62 Coe CL, Kramer M, Czeh B etal. Prenatal PV, Abrous DN. Postnatal stimulation of the
Persistent epigenetic differences associated stress diminishes neurogenesis in the dentate pups counteracts prenatal stress-induced
ho

with prenatal exposure to famine in humans. gyrus of juvenile rhesus monkeys. Biol. deficits in hippocampal neurogenesis. Biol.
Proc. Natl Acad. Sci. USA 105(44), Psychiatry 54(10), 10251034 (2003). Psychiatry 59(9), 786792 (2006).
1704617049 (2008). 63 Fujioka T, Sakata Y, Yamaguchi K, Shibasaki 75 Dicorcia JA, Tronick E. Quotidian resilience:
50 Yehuda R, Bell A, Bierer LM, Schmeidler J. T, Kato H, Nakamura S. The effects of exploring mechanisms that drive resilience
Maternal, not paternal, PTSD is related to prenatal stress on the development of from a perspective of everyday stress and
hypothalamic paraventricular neurons in fetal coping. Neurosci. Biobehav. Rev. 35(7),
ut

increased risk for PTSD in offspring of


Holocaust survivors. J.Psychiatr. Res. 42(13), rats. Neuroscience 92(3), 10791088 (1999). 15931602 (2011).
11041111 (2008). 64 Fujioka A, Fujioka T, Ishida Y, Maekawa T, 76 Francis D, Diorio J, Liu D, Meaney MJ.
51 Yehuda R, Bierer LM. Transgenerational Nakamura S. Differential effects of prenatal Nongenomic transmission across generations
transmission of cortisol and PTSD risk. Prog. stress on the morphological maturation of of maternal behavior and stress responses in
A

Brain Res. 167, 121135 (2008). hippocampal neurons. Neuroscience 141(2), the rat. Science 286(5442), 11551158
907915 (2006). (1999).
52 Francis DD, Meaney MJ. Maternal care and
the development of stress responses. Curr. 65 Fujioka T, Fujioka A, Tan N etal. Mild 77 Champagne FA, Francis DD, Mar A, Meaney
Opin. Neurobiol. 9(1), 128134 (1999). prenatal stress enhances learning performance MJ. Variations in maternal care in the rat as a
in the non-adopted rat offspring. Neuroscience mediating influence for the effects of
53 Wosiski-Kuhn M, Stranahan AM. Opposing
103(2), 301307 (2001). environment on development. Physiol. Behav.
effects of positive and negative stress on
66 Mueller BR, Bale TL. Sex-specific 79(3), 359371 (2003).
hippocampal plasticity over the lifespan.
Ageing Res. Rev. 11(3), 399403 (2012). programming of offspring emotionality after 78 Liu D, Diorio J, Tannenbaum B etal.
stress early in pregnancy. J.Neurosci. 28(36), Maternal care, hippocampal glucocorticoid
54 Wosiski-Kuhn M, Stranahan AM. Opposing
90559065 (2008). receptors, and hypothalamic-pituitary-
effects of positive and negative stress on
67 Morgan CP, Bale TL. Early prenatal stress adrenal responses to stress. Science 277(5332),
hippocampal plasticity over the lifespan.
epigenetically programs dysmasculinization 16591662 (1997).
Ageing Res. Rev. 11(3), 399403 (2011).
in second-generation offspring via the 79 Champagne DL, Bagot RC, Van Hasselt F
55 Koenig JI, Walker CD, Romeo RD, Lupien
paternal lineage. J.Neurosci. 31(33), etal. Maternal care and hippocampal
SJ. Effects of stress across the lifespan. Stress
1174811755 (2011). plasticity. evidence for experience-dependent
14(5), 475480 (2011).
68 Oberlander TF, Weinberg J, Papsdorf M, structural plasticity, altered synaptic
56 Vallee M, Maccari S, Dellu F, Simon H, Le functioning, and differential responsiveness
Grunau R, Misri S, Devlin AM. Prenatal
Moal M, Mayo W. Long-term effects of to glucocorticoids and stress. J.Neurosci.
exposure to maternal depression, neonatal
prenatal stress and postnatal handling on 28(23), 60376045 (2008).
methylation of human glucocorticoid receptor

14 Epigenomics (2013) 5(2) future science group


Stress & anxiety across the lifespan: structural plasticity & epigenetic regulation Review
80 Bagot RC, van Hasselt FN, Champagne DL, transgenerational inheritance of altered stress 105 Roth TL, Lubin FD, Funk AJ, Sweatt JD.
Meaney MJ, Krugers HJ, Joels M. Maternal responses. Proc. Natl Acad. Sci. USA 109(23), Lasting epigenetic influence of early-life
care determines rapid effects of stress 91439148 (2012). adversity on the BDNF gene. Biol. Psychiatry
mediators on synaptic plasticity in adult rat 93 Skinner MK, Anway MD, Savenkova MI, 65(9), 760769 (2009).
hippocampal dentate gyrus. Neurobiol Learn. Gore AC, Crews D. Transgenerational 106 Simmons RK, Howard JL, Simpson DN, Akil
Mem. 92(3), 292300 (2009). epigenetic programming of the brain H, Clinton SM. DNA methylation in the
81 Del Giudice M, Ellis BJ, Shirtcliff EA. The transcriptome and anxiety behavior. PLoS developing hippocampus and amygdala of
Adaptive Calibration Model of stress ONE 3(11), e3745 (2008). anxiety-prone versus risk-taking rats. Dev.
responsivity. Neurosci. Biobehav. Rev. 35(7), 94 Guerrero-Bosagna C, Settles M, Lucker B, Neurosci. 34(1), 5867 (2012).
15621592 (2012). Skinner MK. Epigenetic transgenerational 107 Sturman DA, Moghaddam B. The
82 Weaver IC, Cervoni N, Champagne FA etal. actions of vinclozolin on promoter regions of neurobiology of adolescence: changes in brain
Epigenetic programming by maternal behavior. the sperm epigenome. PLoS ONE 5(9), architecture, functional dynamics, and
Nat. Neurosci. 7(8), 847854 (2004). e13100 (2010). behavioral tendencies. Neurosci. Biobehav. Rev.
83 Cervoni N, Szyf M. Demethylase activity is 95 Dietz DM, Laplant Q, Watts EL etal. 35(8), 17041712 (2011).
directed by histone acetylation. J.Biol. Chem. Paternal transmission of stress-induced 108 Mcgorry PD, Purcell R, Goldstone S,
276(44), 4077840787 (2001). pathologies. Biol. Psychiatry 70(5), 408414 Amminger GP. Age of onset and timing of
84 Weaver IC, Dalessio AC, Brown SE etal. The (2011). treatment for mental and substance use

of
transcription factor nerve growth factor- 96 Walker AK, Hawkins G, Sominsky L, disorders: implications for preventive
inducible protein a mediates epigenetic Hodgson DM. Transgenerational intervention strategies and models of care.
programming: altering epigenetic marks by transmission of anxiety induced by neonatal Curr. Opin. Psychiatry 24(4), 301306 (2011).
immediate-early genes. J.Neurosci. 27(7), exposure to lipopolysaccharide: Implications 109 Blakemore SJ. Development of the social brain

ro
17561768 (2007). for male and female germ lines. in adolescence. J.R.Soc. Med. 105(3), 111116
85 Turner JD, Muller CP. Structure of the Psychoneuroendocrinology 37(8), 13201335 (2012).
glucocorticoid receptor (NR3C1) gene 5 (2012). 110 Gunnar MR, Wewerka S, Frenn K, Long JD,
untranslated region: identification, and tissue 97 Franklin TB, Russig H, Weiss IC etal. Griggs C. Developmental changes in
distribution of multiple new human exon 1. Epigenetic transmission of the impact of early hypothalamus-pituitary-adrenal activity over
rP
J.Mol. Endocrinol. 35(2), 283292 (2005). stress across generations. Biol. Psychiatry the transition to adolescence: normative
86 Mcgowan PO, Sasaki A, Dalessio AC etal. 68(5), 408415 (2010). changes and associations with puberty. Dev.
Epigenetic regulation of the glucocorticoid 98 Liu D, Diorio J, Day JC, Francis DD, Meaney Psychopathol. 21(1), 6985 (2009).
receptor in human brain associates with MJ. Maternal care, hippocampal 111 Romeo RD, Karatsoreos IN, McEwen BS.
childhood abuse. Nat. Neurosci. 12(3), synaptogenesis and cognitive development in Pubertal maturation and time of day
342348 (2009). rats. Nat. Neurosci. 3(8), 799806 (2000). differentially affect behavioral and
ho

87 Labonte B, Yerko V, Gross J etal. Differential 99 Eiland L, Ramroop J, Hill MN, Manley J, neuroendocrine responses following an acute
glucocorticoid receptor exon 1(b), 1(c), and McEwen BS. Chronic juvenile stress produces stressor. Horm. Behav. 50(3), 463468
1(h) expression and methylation in suicide corticolimbic dendritic architectural (2006).
completers with a history of childhood abuse. remodeling and modulates emotional behavior 112 Romeo RD, Bellani R, Karatsoreos IN etal.
Biol. Psychiatry 72(1), 4148 (2012). in male and female rats. Stress history and pubertal development
Psychoneuroendocrinology 37(1), 3947 (2011). interact to shape hypothalamic-pituitary-
ut

88 Alt SR, Turner JD, Klok MD etal. Differential


expression of glucocorticoid receptor 100 Liu D, Caldji C, Sharma S, Plotsky PM, adrenal axis plasticity. Endocrinology 147(4),
transcripts in major depressive disorder is not Meaney MJ. Influence of neonatal rearing 16641674 (2006).
epigenetically programmed. conditions on stress-induced 113 Isgor C, Kabbaj M, Akil H, Watson SJ.
Psychoneuroendocrinology 35(4), 544556 adrenocorticotropin responses and Delayed effects of chronic variable stress
A

(2010). norepinepherine release in the hypothalamic during peripubertal-juvenile period on


89 Tyrka AR, Price LH, Marsit C, Walters OC, paraventricular nucleus. J.Neuroendocrinol. hippocampal morphology and on cognitive
Carpenter LL. Childhood adversity and 12(1), 512 (2000). and stress axis functions in rats. Hippocampus
epigenetic modulation of the leukocyte 101 Murgatroyd C, Patchev AV, Wu Y etal. 14(5), 636648 (2004).
glucocorticoid receptor: preliminary findings Dynamic DNA methylation programs 114 Trollope AF, Gutierrez-Mecinas M, Mifsud
in healthy adults. PLoS ONE 7(1), e30148 persistent adverse effects of early-life stress. KR, Collins A, Saunderson EA, Reul JM.
(2012). Nat. Neurosci. 12(12), 15591566 (2009). Stress, epigenetic control of gene expression
90 Labonte B, Suderman M, Maussion G etal. 102 Kriaucionis S, Bird A. DNA methylation and and memory formation. Exp. Neurol. 233(1),
Genome-wide epigenetic regulation by early- Rett syndrome. Hum. Mol. Genet. 311 (2012).
life traumagenome epigenetic regulation by 12(Suppl.2), R221R227 (2003). 115 Lein ES, Hawrylycz MJ, Ao N etal. Genome-
early-life trauma. Arch. Gen. Psychiatry 69(7), 103 Martinowich K, Hattori D, Wu H etal. DNA wide atlas of gene expression in the adult
722731 (2012). methylation-related chromatin remodeling in mouse brain. Nature 445(7124), 168176
91 Crudo A, Petropoulos S, Moisiadis VG etal. activity-dependent BDNF gene regulation. (2007).
Prenatal synthetic glucocorticoid treatment Science 302(5646), 890893 (2003). 116 Zovkic IB, Sweatt JD. Epigenetic Mechanisms
changes DNA methylation States in male 104 Chen WG, Chang Q, Lin Y etal. in learned fear: implications for PTSD.
organ systems: multigenerational effects. Derepression of BDNF transcription involves Neuropsychopharmacology 38(1), 7793 (2012).
Endocrinology 153(7), 32693283 (2012). calcium-dependent phosphorylation of 117 Sultan FA, Day JJ. Epigenetic mechanisms in
92 Crews D, Gillette R, Scarpino SV, Manikkam MeCP2. Science 302(5646), 885889 (2003). memory and synaptic function. Epigenomics
M, Savenkova MI, Skinner MK. Epigenetic 3(2), 157181 (2011).

future science group www.futuremedicine.com 15


Review Hunter & McEwen

118 Griffith JS, Mahler HR. DNA ticketing glucocorticoid receptor-dependent glucocorticoid receptors with ERK1/2-MSK1-
theory of memory. Nature 223(5206), behavioural response. Eur. J.Neurosci. 22(7), Elk-1 signaling. Proc. Natl Acad. Sci. USA
580582 (1969). 16911700 (2005). 108(33), 1380613811 (2011).
119 Crick F. Memory and molecular turnover. 132 Clayton AL, Rose S, Barratt MJ, Mahadevan 142 Nestler EJ, Hyman SE. Animal models of
Nature 312(5990), 101 (1984). LC. Phosphoacetylation of histone H3 on neuropsychiatric disorders. Nat. Neurosci.
120 Holliday R. Is there an epigenetic component c-fos- and c-jun-associated nucleosomes upon 13(10), 11611169 (2010).
in long-term memory? J.Theor. Biol. 200(3), gene activation. EMBOJ. 19(14), 37143726 143 Tsankova NM, Berton O, Renthal W,
339341 (1999). (2000). KumarA, Neve RL, Nestler EJ. Sustained
121 Levenson JM, Oriordan KJ, Brown KD, 133 Cheung P, Tanner KG, Cheung WL, Sassone- hippocampal chromatin regulation in a
Trinh MA, Molfese DL, Sweatt JD. Corsi P, Denu JM, Allis CD. Synergistic mouse model of depression and
Regulation of histone acetylation during coupling of histone H3 phosphorylation and antidepressant action. Nat. Neurosci. 9(4),
memory formation in the hippocampus. acetylation in response to epidermal growth 519525 (2006).
J.Biol. Chem. 279(39), 4054540559 factor stimulation. Mol. Cell 5(6), 905915 144 Tsankova NM, Kumar A, Nestler EJ. Histone
(2004). (2000). modifications at gene promoter regions in rat
122 Miller CA, Sweatt JD. Covalent modification 134 Crosio C, Heitz E, Allis CD, Borrelli E, hippocampus after acute and chronic
of DNA regulates memory formation. Neuron Sassone-Corsi P. Chromatin remodeling and electroconvulsive seizures. J.Neurosci.
53(6), 857869 (2007). neuronal response: multiple signaling 24(24), 56035610 (2004).

of
pathways induce specific histone H3 145 Renthal W, Maze I, Krishnan V etal.
123 Lubin FD, Roth TL, Sweatt JD. Epigenetic
modifications and early gene expression in Histone deacetylase 5 epigenetically controls
regulation of BDNF gene transcription in the
hippocampal neurons. J.Cell Sci. 116(Pt 24), behavioral adaptations to chronic emotional
consolidation of fear memory. J.Neurosci.
49054914 (2003). stimuli. Neuron 56(3), 517529 (2007).
28(42), 1057610586 (2008).

ro
135 Chandramohan Y, Droste SK, Arthur JS, 146 Covington HE, 3rd, Maze I, Laplant QC
124 Koshibu K, Graff J, Beullens M etal. Protein
Reul JM. The forced swimming-induced etal. Antidepressant actions of histone
phosphatase 1 regulates the histone code for
behavioural immobility response involves deacetylase inhibitors. J.Neurosci. 29(37),
long-term memory. J.Neurosci. 29(41),
histone H3 phospho-acetylation and c-Fos 1145111460 (2009).
1307913089 (2009).
induction in dentate gyrus granule neurons
147 Laplant Q, Vialou V, Covington HE 3rd etal.
rP
125 Koshibu K, Graff J, Mansuy IM. Nuclear via activation of the N-methyl-d-aspartate/
protein phosphatase-1: an epigenetic regulator Dnmt3a regulates emotional behavior and
extracellular signal-regulated kinase/mitogen-
of fear memory and amygdala long-term spine plasticity in the nucleus accumbens.
and stress-activated kinase signalling pathway.
potentiation. Neuroscience 173, 3036 (2011). Nat. Neurosci. 13(9), 11371143 (2010).
Eur. J.Neurosci. 27(10), 27012713 (2008).
126 Maddox SA, Schafe GE. Epigenetic 148 Elliott E, Ezra-Nevo G, Regev L, Neufeld-
136 Chandramohan Y, Droste SK, Reul JM.
alterations in the lateral amygdala are Cohen A, Chen A. Resilience to social stress
Novelty stress induces phospho-acetylation of
coincides with functional DNA methylation
ho

required for reconsolidation of a Pavlovian histone H3 in rat dentate gyrus granule


fear memory. Learn. Mem. 18(9), 579593 of the Crf gene in adult mice. Nat. Neurosci.
neurons through coincident signalling via the
(2011). 13(11), 13511353 (2010).
N-methyl-D-aspartate receptor and the
127 Fischer A, Sananbenesi F, Wang X, glucocorticoid receptor: relevance for c-fos 149 Wilkinson MB, Xiao G, Kumar A etal.
DobbinM, Tsai LH. Recovery of learning induction. J.Neurochem. 101(3), 815828 Imipramine treatment and resiliency exhibit
and memory is associated with chromatin (2007). similar chromatin regulation in the mouse
ut

remodelling. Nature 447(7141), 178182 nucleus accumbens in depression models.


137 Collins A, Hill LE, Chandramohan Y,
(2007). J.Neurosci. 29(24), 78207832 (2009).
Whitcomb D, Droste SK, Reul JM. Exercise
128 Guan JS, Haggarty SJ, Giacometti E etal. improves cognitive responses to psychological 150 Covington HE, 3rd, Maze I, Sun H etal. A
HDAC2 negatively regulates memory stress through enhancement of epigenetic role for repressive histone methylation in
mechanisms and gene expression in the cocaine-induced vulnerability to stress.
A

formation and synaptic plasticity. Nature


459(7243), 5560 (2009). dentate gyrus. PLoS ONE 4(1), e4330 (2009). Neuron 71(4), 656670 (2011).

129 Bredy TW, Wu H, Crego C, Zellhoefer J, 138 Papadopoulos A, Chandramohan Y, Collins 151 Maze I, Covington HE 3rd, Dietz DM etal.
Sun YE, Barad M. Histone modifications A, Droste SK, Nutt DJ, Reul JM. GABAergic Essential role of the histone methyltransferase
around individual BDNF gene promoters in control of novelty stress-responsive epigenetic G9a in cocaine-induced plasticity. Science
prefrontal cortex are associated with and gene expression mechanisms in the rat 327(5962), 213216 (2010).
extinction of conditioned fear. Learn. Mem. dentate gyrus. Eur. Neuropsychopharmacol. 152 Hunter RG, Mccarthy KJ, Milne TA,
14(4), 268276 (2007). 21(4), 316324 (2011). PfaffDW, McEwen BS. Regulation of
130 Calfa G, Chapleau CA, Campbell S etal. 139 McEwen BS. Stress and hippocampal hippocampal H3 histone methylation by acute
HDAC activity is required for BDNF to plasticity. Annu. Rev. Neurosci. 22, 105122 and chronic stress. Proc. Natl Acad. Sci. USA
increase quantal neurotransmitter release and (1999). 106(49), 2091220917 (2009).
dendritic spine density in CA1 pyramidal 140 Reul JM, Hesketh SA, Collins A, Mecinas 153 Reed B, Fang N, Mayer-Blackwell B etal.
neurons. Hippocampus 22(7), 14931500 MG. Epigenetic mechanisms in the dentate Chromatin alterations in response to forced
(2011). gyrus act as a molecular switch in swimming underlie increased prodynorphin
131 Bilang-Bleuel A, Ulbricht S, Chandramohan hippocampus-associated memory formation. transcription. Neuroscience 220, 109118
Y, De Carli S, Droste SK, Reul JM. Epigenetics 4(7), 434439 (2009). (2012).
Psychological stress increases histone H3 141 Gutierrez-Mecinas M, Trollope AF, Collins A 154 Mcclintock B. The significance of responses of
phosphorylation in adult dentate gyrus etal. Long-lasting behavioral responses to the genome to challenge. Science 226(4676),
granule neurons: involvement in a stress involve a direct interaction of 792801 (1984).

16 Epigenomics (2013) 5(2) future science group


Stress & anxiety across the lifespan: structural plasticity & epigenetic regulation Review
155 Hunter RG, Murakami G, Dewell S etal. 168 Sanosaka T, Namihira M, Nakashima K. from Alzheimers disease and bipolar disorder
Acute stress and hippocampal histone H3 Epigenetic mechanisms in sequential patients. Transl. Psychiatry 2, e132 (2012).
lysine 9 trimethylation, a retrotransposon differentiation of neural stem cells. Epigenetics 183 Anderson RM, Shanmuganayagam D,
silencing response. Proc. Natl Acad. Sci. USA 4(2), 8992 (2009). Weindruch R. Caloric restriction and aging:
109(43), 1765717662 (2012). 169 Sapolsky RM, Krey LC, McEwen BS. studies in mice and monkeys. Toxicol. Pathol.
156 Rusiecki JA, Chen L, Srikantan V etal. DNA Corticosterone receptors decline in a site- 37(1), 4751 (2009).
methylation in repetitive elements and post- specific manner in the aged rat brain. Brain 184 Chouliaras L, Van Den Hove DL, Kenis G
traumatic stress disorder: a case-control study Res. 289(12), 235240 (1983). etal. Caloric restriction attenuates age-related
of US military service members. Epigenomics 170 Sapolsky RM, Krey LC, McEwen BS. The changes of DNA methyltransferase 3a in
4(1), 2940 (2012). adrenocortical stress-response in the aged mouse hippocampus. Brain Behav. Immun.
157 Hedges DJ, Belancio VP. Restless genomes male rat: impairment of recovery from stress. 25(4), 616623 (2010).
humans as a model organism for Exp. Gerontol. 18(1), 5564 (1983). 185 Patel NV, Finch CE. The glucocorticoid
understanding host-retrotransposable element 171 Sapolsky RM, Krey LC, McEwen BS. The paradox of caloric restriction in slowing brain
dynamics. Adv. Genet. 73, 219262 (2011). neuroendocrinology of stress and aging: the aging. Neurobiol. Aging 23(5), 707717
158 Uchida S, Nishida A, Hara K etal. glucocorticoid cascade hypothesis. Endocr. (2002).
Characterization of the vulnerability to Rev. 7(3), 284301 (1986). 186 McEwen BS. Plasticity of the hippocampus:
repeated stress in Fischer 344 rats: possible 172 Garrido P. Aging and stress: past hypotheses, adaptation to chronic stress and allostatic

of
involvement of microRNA-mediated down- present approaches and perspectives. Aging load. Ann. NY Acad. Sci. 933, 265277
regulation of the glucocorticoid receptor. Eur. Dis 2(1), 8099 (2012). (2001).
J.Neurosci. 27(9), 22502261 (2008).
173 Lupien S, Lecours AR, Schwartz G etal. 187 Szulwach KE, Li X, Li Y etal. 5-hmC-
159 Vreugdenhil E, Verissimo CS, Mariman R Longitudinal study of basal cortisol levels in mediated epigenetic dynamics during

ro
etal. MicroRNA 18 and 124a down-regulate healthy elderly subjects: evidence for postnatal neurodevelopment and aging. Nat.
the glucocorticoid receptor: implications for subgroups. Neurobiol. Aging 17(1), 95105 Neurosci. 14(12), 16071616 (2011).
glucocorticoid responsiveness in the brain. (1996). 188 Chen H, Dzitoyeva S, Manev H. Effect of
Endocrinology 150(5), 22202228 (2009).
174 Foy MR, Baudry M, Foy JG, Thompson RF. aging on 5-hydroxymethylcytosine in the
160 Meerson A, Cacheaux L, Goosens KA, 17beta-estradiol modifies stress-induced and mouse hippocampus. Restor. Neurol. Neurosci.
rP
Sapolsky RM, Soreq H, Kaufer D. Changes age-related changes in hippocampal synaptic 30(3), 237245 (2012).
in brain MicroRNAs contribute to cholinergic plasticity. Behav. Neurosci. 122(2), 301309 189 Graff J, Rei D, Guan JS etal. An epigenetic
stress reactions. J.Mol. Neurosci. 40(12), (2008). blockade of cognitive functions in the
4755 (2010).
175 Lee SW, Clemenson GD, Gage FH. New neurodegenerating brain. Nature 483(7388),
161 Haramati S, Navon I, Issler O etal. neurons in an aged brain. Behav. Brain Res. 222226 (2012).
MicroRNA as repressors of stress-induced 227(2), 497507 (2012).
ho

190 Zeng Y, Tan M, Kohyama J etal. Epigenetic


anxiety: the case of amygdalar miR-34.
176 Burke SN, Barnes CA. Senescent synapses enhancement of BDNF signaling rescues
J.Neurosci. 31(40), 1419114203 (2011).
and hippocampal circuit dynamics. Trends synaptic plasticity in aging. J.Neurosci.
162 Rinaldi A, Vincenti S, De Vito F etal. Stress Neurosci. 33(3), 153161 (2010). 31(49), 1780017810 (2011).
induces region specific alterations in
177 Heyn H, Li N, Ferreira HJ etal. Distinct 191 Peleg S, Sananbenesi F, Zovoilis A etal.
microRNAs expression in mice. Behav. Brain
DNA methylomes of newborns and Altered histone acetylation is associated with
Res. 208(1), 265269 (2010).
ut

centenarians. Proc. Natl Acad. Sci. USA age-dependent memory impairment in mice.
163 Babenko O, Golubov A, Ilnytskyy Y, 109(26), 1052210527 (2012). Science 328(5979), 753756 (2010).
Kovalchuk I, Metz GA. Genomic and
178 Dosunmu R, Alashwal H, Zawia NH. 192 Walker MP, Laferla FM, Oddo SS, Brewer
Epigenomic responses to chronic stress
Genome-wide expression and methylation GJ. Reversible epigenetic histone
involve miRNA-mediated programming.
A

profiling in the aged rodent brain due to modifications and Bdnf expression in neurons
PLoS ONE 7(1), e29441 (2012).
early-life Pb exposure and its relevance to with aging and from a mouse model of
164 Mongrain V, Hernandez SA, Pradervand S aging. Mech. Ageing Dev. 133(6), 435443 Alzheimers disease. Age (Dordr.) doi:10.1007/
etal. Separating the contribution of (2012). s11357-011-9375-5 (2012) (Epub ahead of
glucocorticoids and wakefulness to the print).
179 Thompson RF, Atzmon G, Gheorghe C etal.
molecular and electrophysiological correlates
Tissue-specific dysregulation of DNA 193 Castellano JF, Fletcher BR, Kelley-Bell B,
of sleep homeostasis. Sleep 33(9), 11471157
methylation in aging. Aging Cell 9(4), Kim DH, Gallagher M, Rapp PR. Age-related
(2010).
506518 (2010). memory impairment is associated with
165 Rajasethupathy P, Antonov I, Sheridan R disrupted multivariate epigenetic
180 Hernandez DG, Nalls MA, Gibbs JR etal.
etal. A role for neuronal piRNAs in the coordination in the hippocampus. PLoS ONE
Distinct DNA methylation changes highly
epigenetic control of memory-related synaptic 7(3), e33249 (2012).
correlated with chronological age in the
plasticity. Cell 149(3), 693707 (2012).
human brain. Hum. Mol. Genet. 20(6), 194 Doyle JP, Dougherty JD, Heiman M etal.
166 Mccue AD, Nuthikattu S, Reeder SH, Slotkin 11641172 (2011). Application of a translational profiling
RK. Gene expression and stress response approach for the comparative analysis of CNS
181 Numata S, Ye T, Hyde TM etal. DNA
mediated by the epigenetic regulation of a cell types. Cell 135(4), 749762 (2008).
methylation signatures in development and
transposable element small RNA. PLoS Genet.
aging of the human prefrontal cortex. Am. 195 Heiman M, Schaefer A, Gong S etal. A
8(2), e1002474 (2012).
J.Hum. Genet. 90(2), 260272 (2012). translational profiling approach for the
167 Perdiguero E, Sousa-Victor P, Ballestar E, molecular characterization of CNS cell types.
182 Rao JS, Keleshian VL, Klein S, Rapoport SI.
Munoz-Canoves P. Epigenetic regulation of Cell 135(4), 738748 (2008).
Epigenetic modifications in frontal cortex
myogenesis. Epigenetics 4(8), 541550 (2009).

future science group www.futuremedicine.com 17


Review Hunter & McEwen

196 Veenema AH. Early life stress, the 198 Zhang D, Yu ZY, Cruz P, Kong Q, Li S, Kone 201 Kubicek S, OSullivan RJ, August EM etal.
development of aggression and BC. Epigenetics and the control of epithelial Reversal of H3K9me2 by a small-molecule
neuroendocrine and neurobiological sodium channel expression in collecting duct. inhibitor for the G9a histone
correlates: what can we learn from animal Kidney Int. 75(3), 260267 (2009). methyltransferase. Mol. Cell 25(3), 473481
models? Front. Neuroendocrinol. 30(4), 199 ENCODE Project Consortium. The (2007).
497518 (2009). ENCODE (ENCyclopedia Of DNA 202 Hunter RG. Epigenetic effects of stress and
197 Kolber BJ, Wieczorek L, Muglia LJ. Elements) Project. Science 306(5696), corticosteroids in the brain. Front. Cell.
Hypothalamic-pituitary-adrenal axis 636640 (2004). Neurosci. 6, 18 (2012).
dysregulation and behavioral analysis of 200 Spannhoff A, Hauser AT, Heinke R, Sippl W, 203 Magarinos AM, McEwen BS, Flugge G,
mouse mutants with altered glucocorticoid or Jung M. The emerging therapeutic potential Fuchs E. Chronic psychosocial stress causes
mineralocorticoid receptor function. Stress of histone methyltransferase and demethylase apical dendritic atrophy of hippocampal CA3
11(5), 321338 (2008). inhibitors. ChemMedChem. 4(10), 15681582 pyramidal neurons in subordinate tree shrews.
(2009). J.Neurosci. 16(10), 35343540 (1996).

of
ro
rP
ho
ut
A

18 Epigenomics (2013) 5(2) future science group

Das könnte Ihnen auch gefallen